Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Toxicol Appl Pharmacol ; 449: 116139, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35750203

ABSTRACT

Biomacromolecules such as proteins and nucleic acids are very attractive due to their high efficiency and specificity as cancer therapeutics. In fact, the endocytosed macromolecules are often trapped in the endosomes and cannot exhibit pharmacological effects well. Many strategies have been used to address this bottleneck, and one promising approach is to exploit the endosomal escape-promoting effect of triterpenoid saponins to aid in the release of biomacromolecules. Here, Raddeanin A (RA, an oleanane-type triterpenoid saponin) was proved to significantly promote endosomal escape as it recruited Galectin-9, an endosomal escape event reporter. As expected, RA effectively enhanced the anti-tumor effect of MAP30 (a type I ribosome-inactivating protein derived from Momordica charantia). However, based on the results of fluorescent colocalization, RA did not significantly promote MAP30 release from endosomes, suggesting that RA enhances MAP30 activity not only by promoting endosomal escape. Furthermore, it was found that the inhibitors of micropinocytosis and caveolae could almost completely inhibit the cytotoxicity of MAP30 combined with RA without affecting the cytotoxicity of MAP30 alone, indicating that RA may regulate the endocytic pathway of MAP30. Meanwhile, the effect of RA is related to the intra vesicular pH and cholesterol content on cell membrane, and is also cell-type dependent. Therefore, RA enhanced the anti-tumor effect of MAP30 in multiple ways, not just by promoting endosomal escape. Our findings will help to further decipher the possible mechanisms by which triterpenoid saponins enhance drug activity, and provide a new perspective for improving the activity of endocytosed drugs.


Subject(s)
Neoplasms , Saponins , Triterpenes , Endosomes/metabolism , Humans , Neoplasms/metabolism , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/pharmacology , Saponins/pharmacology , Triterpenes/pharmacology
2.
Protein Expr Purif ; 185: 105893, 2021 09.
Article in English | MEDLINE | ID: mdl-33933613

ABSTRACT

MAP30 (Momordica antiviral protein 30kD) is a single-chain Ⅰ-type ribosome inactivating protein with a variety of biological activities, including anti-tumor ability. It was reported that MAP30 would serve as a novel and relatively safe agent for prophylaxis and treatment of liver cancer. To determine whether adding two tumor targeting peptides could improve the antitumor activities of MAP30, we genetically modified MAP30 with an RGD motif and a EGFRi motif, which is a ligand with high affinity for αvß3 integrins and with high affinity for EGFR. The recombinant protein ELRL-MAP30 (rELRL-MAP30) containing a GST-tag was expressed in E. coli. The rELRL-MAP30 was highly expressed in the soluble fraction after induction with 0.15 mM IPTG for 20 h at 16 °C. The purified rELRL-MAP30 appeared as a band on SDS-PAGE. It was identified by western blotting. Cytotoxicity of recombinant protein to HepG2, MDA-MB-231, HUVEC and MCF-7 cells was detected by MTT analysis. Half maximal inhibitory concentration (IC50) values were 54.64 µg/mL, 70.13 µg/mL, 146 µg/mL, 466.4 µg/mL, respectively. Proliferation inhibition assays indicated that rELRL-MAP30 could inhibit the growth of Human liver cancer cell HepG2 effectively. We found that rELRL-MAP30 significantly induced apoptosis in liver cancer cells, as evidenced by nuclear staining of DAPI. In addition, rELRL-MAP30 induced apoptosis in human liver cancer HepG2 cells by up-regulation of Bax as well as down-regulation of Bcl-2. Migration of cell line were markedly inhibited by rELRL-MAP30 in a dose-dependent manner compared to the recombinant MAP30 (rMAP30). In summary, the fusion protein displaying extremely potent cytotoxicity might be highly effective for tumor therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Momordica charantia/chemistry , Peptides/genetics , Recombinant Fusion Proteins/genetics , Ribosome Inactivating Proteins, Type 2/genetics , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cloning, Molecular , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha5/genetics , Integrin alpha5/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , MCF-7 Cells , Peptides/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
3.
Pharmacol Res ; 161: 105157, 2020 11.
Article in English | MEDLINE | ID: mdl-32814169

ABSTRACT

Increasing evidence shows that Traditional Chinese Medicine (TCM) has an obvious appeal for cancer treatment, but there is still a lack of scientific investigation of its underlying molecular mechanisms. Bitter melon or bitter gourd (Momordica charantia) is an edible fruit that is commonly consumed, and it is used to cure different diseases in various ancient folk medical practices. We report that a bioactive protein, MAP30, isolated from bitter melon seeds exhibited potent anticancer and anti-chemoresistant effects on ovarian cancer cells. Functional studies revealed that MAP30 inhibited cancer cell migration, cell invasion, and cell proliferation in various ovarian cancer cells but not normal immortalized ovarian epithelial cells. When administered with cisplatin, MAP30 produced a synergistic effect on cisplatin-induced cell cytotoxicity in ovarian cancer cells. When low doses of cisplatin and MAP30 were co-injected intraperitoneally, a remarkable reduction of tumor dissemination and tumor growth was observed in an ovarian cancer ascites mouse model. Notably, blood tests confirmed that MAP30 did not cause any adverse effects on liver and kidney functions in the treated mice. MAP30 activated AMP-activated protein kinase (AMPK) signaling via CaMKKß and induced cell cycle arrest in the S-phase. MAP30 modulated cell metabolism of ovarian cancer cells via suppression of GLUT-1/-3-mediated glucose uptake, adipogenesis, and lipid droplet formation in tumor development and progression. MAP30 also induced an increase in intracellular Ca2+ ion concentration, which triggered ROS-mediated cancer cell death via apoptosis and ferroptosis. Collectively, these findings suggest that natural MAP30 is a non-toxic supplement that may enhance chemotherapeutic outcomes and benefit ovarian cancer patients with peritoneal metastases.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , Energy Metabolism/drug effects , Ferroptosis/drug effects , Momordica charantia , Ovarian Neoplasms/drug therapy , Ribosome Inactivating Proteins, Type 2/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Female , Glycolysis/drug effects , Humans , Lipogenesis/drug effects , Mice, Inbred BALB C , Mice, Nude , Momordica charantia/chemistry , Neoplasm Invasiveness , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ribosome Inactivating Proteins, Type 2/isolation & purification , Xenograft Model Antitumor Assays
4.
Dokl Biochem Biophys ; 494(1): 219-221, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33119820

ABSTRACT

In the present study, we assessed the role of annexin 13 membrane-binding protein (ANXA13) in the intracellular transport of vesicles containing type II ribosome-inactivating proteins (RIP-IIs). A modified human intestinal epithelial cell line HT29 was used, in which the expression of ANXA13 was significantly reduced. The cytotoxic effect of ricin and viscumin was evaluated by modification of 28S ribosome RNA. The observed differences in the activity of toxins on the parental and modified HT29 lines indicate that ANXA13 plays a different role in the intracellular transport of vesicles containing the RIP-IIs.


Subject(s)
Annexins/metabolism , Chemical Warfare Agents/pharmacology , Colonic Neoplasms/pathology , Ribosome Inactivating Proteins, Type 2/pharmacology , Ribosome Inactivating Proteins/metabolism , Ribosomes/drug effects , Ricin/pharmacology , Toxins, Biological/pharmacology , Biological Transport , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , HT29 Cells , Humans
5.
Dokl Biochem Biophys ; 493(1): 198-200, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32894464

ABSTRACT

The role of proteasome proteins and proteins of the ERAD system in the cytotoxicity of type II ribosome-inactivating proteins ricin and viscumin was investigated. For this, the cell line of colorectal adenocarcinoma HT29, as well as the HT29-sh002 line obtained on its basis, were used. On the basis on the proteome analysis of these lines and the estimation of the proportion of inactivated ribosomes, it was shown that the contribution of the proteasome to the degradation of the catalytic subunits of toxins is different. The role of the Cdc37 co-chaperone in maintaining the stability of A subunit of viscumin in the cytoplasm is shown.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , Colorectal Neoplasms/drug therapy , Proteasome Endopeptidase Complex/biosynthesis , Ribosome Inactivating Proteins, Type 2/pharmacology , Ricin/pharmacology , Toxins, Biological/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Cycle Proteins/genetics , Chaperonins/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Humans , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Ribosomes/metabolism , Tumor Cells, Cultured
6.
Anticancer Drugs ; 29(8): 736-747, 2018 09.
Article in English | MEDLINE | ID: mdl-29912010

ABSTRACT

Cyclosporin A (CsA) is a calcium antagonist and can enhance the efficacy of some protein drugs, but its mechanism remains unknown. In this study, MAP30, a ribosome-inactivating protein reported to have apoptotic effects on cancer cells, was fused with S3, an epidermal growth factor receptor (EGFR)-targeting peptide. In addition, CsA was used to investigate whether it can further promote the apoptotic effects of S3 fused MAP30 (MAP30-S3). Our result showed that the internalization of FITC-labeled MAP30-S3 was increased significantly by S3 in HeLa cells. Unexpectedly, MAP30-S3 only showed a minor decrease in the viability of EGFR-overexpressing cancer cells, including HeLa, SMMC-7721, and MGC803 (IC50>5 µmol/l). However, 2 µmol/l CsA significantly increased the cytotoxicity of MAP30-S3, especially for HeLa cells (IC50=40.3 nmol/l). In comparison, CsA did not further decrease the cytotoxicity of MAP30-S3 on MRC-5, an EGFR low-expressing cell line from normal lung tissue, indicating that CsA did not affect the cancer-targeting specificity of MAP30-S3. Our results also showed that CsA further increased the apoptotic activity of MAP30-S3 in HeLa cells. CsA could promote the endosomal escape of FITC-MAP30-S3 with a diffused pattern in the cytoplasm. Five endocytic inhibitors were used to investigate the cellular uptake mechanism of MAP30-S3, and the results showed that the endosomal escape-enhancing effect of CsA on MAP30-S3 may be associated with the clathrin-dependent endocytic pathways. Our study suggested that CsA could be a novel endosomal escape enhancer to potentiate the intracellular release of anticancer protein drugs, resulting in their improved therapeutic efficacy.


Subject(s)
Cyclosporine/pharmacology , Endosomes/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Recombinant Fusion Proteins/pharmacology , Ribosomal Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Synergism , HeLa Cells , Humans , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/genetics
7.
Biochim Biophys Acta Gen Subj ; 1862(3): 460-473, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29154940

ABSTRACT

BACKGROUND: Sambucus ebulus is a rich source of ribosome-inactivating proteins (RIPs) and RIP-related lectins generated from multiple genes. These proteins differ in their structure, enzymatic activity and sugar binding specificity. METHODS: We have purified and characterized ebulin-RP from S. ebulus leaves and determined the amino acid sequence by cDNA cloning. Cytotoxicity was studied in a variety of cancer cells and a comparative study of the ability of ebulin-RP to bind sugars using "in vitro" and "in silico" approaches was performed. RESULTS: Ebulin-RP is a novel heterodimeric type 2 RIP present in S. ebulus leaves together with the type 2 RIP ebulin l, which displayed rRNA N-glycosidase activity but unlike ebulin l, lacked functional sugar binding domains. As a consequence of changes in its B-chain, ebulin-RP displayed lower cytotoxicity than ebulin l towards cancer cells and induced apoptosis as the predominant pattern of cell death. CONCLUSIONS: Ebulin-RP is a novel member of the ebulin gene family with low cytotoxicity as a result of deficient sugar binding domains. Type 2 RIP genes from Sambucus have evolved to render proteins with different sugar affinities that may be related to different biological activities and could result in an advantage for the plant. GENERAL SIGNIFICANCE: The ebulin family of RIPs and lectins can serve as a good model for studying the evolutionary process which may have occurred in RIPs. The lack of cytotoxicity of ebulin-RP makes it a good candidate as a toxic moiety in the construction of immunotoxins and conjugates directed against specific targets.


Subject(s)
Cytotoxins/isolation & purification , Ribosome Inactivating Proteins, Type 2/isolation & purification , Sambucus/enzymology , Sugars/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Binding Sites , Cell Line , Cell Line, Tumor , Cell-Free System , Cytotoxins/chemistry , Cytotoxins/metabolism , Cytotoxins/pharmacology , Drug Screening Assays, Antitumor , Evolution, Molecular , Humans , Mesenchymal Stem Cells/drug effects , Mice , Models, Molecular , Molecular Docking Simulation , Nucleic Acids/drug effects , Phylogeny , Plant Leaves/enzymology , Protein Conformation , Protein Domains , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
8.
Mol Biol (Mosk) ; 52(4): 675-682, 2018.
Article in Russian | MEDLINE | ID: mdl-30113033

ABSTRACT

The mistletoe lectin viscumin (MLI) is a ribosome-inactivating protein from Viscum album widely used in cancer therapy. Its antitumor properties are due to its immunomodulating action, previously demonstrated in experiments involving intravenous, subcutaneous, and oral administration of viscumin. To investigate whether viscumin has a cytotoxic effect on the intestinal epithelium, its safety was assessed using (i) impedance spectroscopy to measure the integrity of the colorectal adenocarcinoma Caco-2 cell monolayer after exposure to viscumin and (ii) a novel technique of determining the portion of viscumin-inactivated ribosomes. It was shown that inactivation of at least 20% of the ribosomes within 6 h did not lead to disruption of the Caco-2 cell monolayer or alter the physicochemical parameters of enterocyte membranes.


Subject(s)
Colorectal Neoplasms/drug therapy , Intestinal Mucosa/drug effects , Ribosome Inactivating Proteins, Type 2/pharmacology , Ribosomes/drug effects , Toxins, Biological/pharmacology , Caco-2 Cells , Cell Membrane/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/pathology , Electric Impedance , Enterocytes/drug effects , Humans , Ribosomes/genetics
9.
Mol Carcinog ; 56(2): 389-401, 2017 02.
Article in English | MEDLINE | ID: mdl-27182794

ABSTRACT

Abrus agglutinin (AGG), a type II ribosome-inactivating protein has been found to induce mitochondrial apoptosis. In the present study, we documented that AGG-mediated Akt dephosphorylation led to ER stress resulting the induction of autophagy-dependent cell death through the canonical pathway in cervical cancer cells. Inhibition of autophagic death with 3-methyladenine (3-MA) and siRNA of Beclin-1 and ATG5 increased AGG-induced apoptosis. Further, inhibiting apoptosis by Z-DEVD-FMK and N-acetyl cysteine (NAC) increased autophagic cell death after AGG treatment, suggesting that AGG simultaneously induced autophagic and apoptotic death in HeLa cells. Additionally, it observed that AGG-induced autophagic cell death in Bax knock down (Bax-KD) and 5-FU resistant HeLa cells, confirming as an alternate cell killing pathway to apoptosis. At the molecular level, AGG-induced ER stress in PERK dependent pathway and inhibition of ER stress by salubrinal, eIF2α phosphatase inhibitor as well as siPERK reduced autophagic death in the presence of AGG. Further, our in silico and colocalization study showed that AGG interacted with pleckstrin homology (PH) domain of Akt to suppress its phosphorylation and consequent downstream mTOR dephosphorylation in HeLa cells. We showed that Akt overexpression could not augment GRP78 expression and reduced autophagic cell death by AGG as compared to pcDNA control, indicating Akt modulation was the upstream signal during AGG's ER stress mediated autophagic cell death. In conclusion, we established that AGG stimulated cell death by autophagy might be used as an alternative tumor suppressor mechanism in human cervical cancer. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Plant Lectins/pharmacology , Pleckstrin Homology Domains/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , Abrus/chemistry , Antineoplastic Agents/isolation & purification , Endoplasmic Reticulum Chaperone BiP , Female , HeLa Cells , Humans , Models, Molecular , Plant Lectins/isolation & purification , Proto-Oncogene Proteins c-akt/chemistry , Ribosome Inactivating Proteins, Type 2/isolation & purification , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , eIF-2 Kinase/metabolism
10.
Mol Cell Biochem ; 426(1-2): 87-99, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27868169

ABSTRACT

Leukemia is among the most aggressive and prevalent human malignant carcinoma. Chemotherapy is the preferred therapeutic strategy; however, recurrence of cancer and non-selective cytotoxicity are the major concerns. Unlike synthetic chemotherapeutic agents, mistletoe ribosome-inactivating protein (RIP) displays anti-tumor function in various types of cancers. However, its effect on leukemia cells is little explored. In this study, we assessed the impact of Viscum articulatum RIP (Articulatin-D) on the survival of acute T-cell leukemia cells and the involved molecular and cellular mechanisms. Cell proliferation assay showed that Articulatin-D suppressed the viability of leukemia cells selectively. We further confirmed that the elevation of mitochondrial membrane potential and exposure of phosphatidylserine are the early events of apoptosis induction in Articulatin-D-treated Jurkat cells. Subsequently, we found that Articulatin-D treatment induces apoptosis in Jurkat cells in a time- and concentration-dependent manner. In conclusion, we provided evidence that Articulatin-D efficiently activates caspase-8 involved in extrinsic pathway of apoptosis induction, which ultimately results in caspase-3-dependent DNA fragmentation of Jurkat cells. Further evaluation of Articulatin-D in cell culture and animal models may provide novel information on selective cytotoxicity to acute T-cell leukemia and its involvement in targeting tumor cell survival pathways.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Cell Proliferation/drug effects , Plant Preparations/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Ribosome Inactivating Proteins, Type 2/pharmacology , Toxins, Biological/pharmacology , Viscum/chemistry , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , Humans , Jurkat Cells , Plant Preparations/chemistry , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Ribosome Inactivating Proteins, Type 2/chemistry , Toxins, Biological/chemistry
12.
Bull Exp Biol Med ; 163(6): 745-748, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29063321

ABSTRACT

External magnetic field is characterized by low toxicity and existence of magnetic properties, which contributes to an interest in the development of products from ferromagnetic nanoparticles (FNP) for antitumor therapy. Previously we synthesized a conjugate of ferromagnetic magnetite nanoparticles and viscumin (mistletoe lectin I, MLI), which exhibits the antitumor activity. Studying the pharmacological properties of this conjugate (FNP-MLI) was directed to the evaluation of FNP-MLI elimination after intratumor injection in mice. The elimination rate of FNP-MLI was much lower than that of native plant MLI. The presence of FNP-MLI was not accompanied by undesired changes in the tumor tissue. The use of a FNP-MLI conjugate allowed us to prolong the time of MLI presence in tissues without increasing the dose of exogenous lectin. These features contribute to the prolongation of an immunomodulatory effect of MLI.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacokinetics , Breast Neoplasms/drug therapy , Drug Carriers/administration & dosage , Magnetite Nanoparticles/administration & dosage , Ribosome Inactivating Proteins, Type 2/pharmacokinetics , Toxins, Biological/pharmacokinetics , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Carriers/pharmacokinetics , Female , Humans , Injections, Intralesional , Injections, Subcutaneous , Magnetic Resonance Imaging , Mice , Mice, SCID , Ribosome Inactivating Proteins, Type 2/pharmacology , Toxins, Biological/pharmacology , Xenograft Model Antitumor Assays
13.
J Pept Sci ; 22(11-12): 689-699, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27739168

ABSTRACT

Cell-penetrating peptides (CPPs) have been shown to be potential drug carriers for cancer therapy. The inherently low immunogenicity and cytotoxicity of human-derived CPPs make them more suitable for intracellular drug delivery compared to other delivery vehicles. In this work, the protein transduction ability of a novel CPP (termed HBP) derived from the heparin-binding domain of HB-EGF was evaluated. Our data shows, for the first time, that HBP possesses similar properties to typical CPPs and is a potent drug delivery vector for improving the antitumor activity of impermeable MAP30. The intrinsic bioactivities of recombinant MAP30-HBP were well preserved compared to those of free MAP30. Furthermore, HBP conjugated to the C-terminus of MAP30 promoted the cellular uptake of recombinant MAP30-HBP. Moreover, the fusion of HBP to MAP30 gave rise to significantly enhanced cytotoxic effects in all of the tumor cell lines tested. In HeLa cells, this cytotoxicity was mainly caused by the induction of cell apoptosis. Further investigation revealed that HBP enhanced MAP30-induced apoptosis through the activation of the mitochondrial- and death receptor-mediated signaling pathways. In addition, the MAP30-HBP fusion protein caused more HeLa cells to become arrested in S phase compared to MAP30 alone. These results highlight the MAP30-HBP fusion protein as a promising drug candidate for cancer therapy and demonstrate HBP, a novel CPP derived from human HB-EGF, as a new potential vector for antitumor drug delivery. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Drug Carriers/pharmacology , Heparin-binding EGF-like Growth Factor/pharmacology , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/pharmacology , Amino Acid Sequence , Apoptosis/drug effects , Cell Line, Tumor , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/genetics , Cloning, Molecular , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Carriers/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Heparin/chemistry , Heparin/metabolism , Heparin-binding EGF-like Growth Factor/biosynthesis , Heparin-binding EGF-like Growth Factor/genetics , Humans , Momordica charantia/chemistry , Protein Binding , Protein Domains , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Ribosome Inactivating Proteins, Type 2/biosynthesis , Ribosome Inactivating Proteins, Type 2/genetics , S Phase/drug effects , Signal Transduction
14.
Molecules ; 21(11)2016 Oct 31.
Article in English | MEDLINE | ID: mdl-27809255

ABSTRACT

PEGylation is a well-established and effective strategy to decrease immunogenicity, which can increase the stability and in vivo half-life time. However, the generation of multi-site modified products is inevitable due to the lysine chemistry, which will bring difficulties in subsequent research, such as purification and quantification. Site-specific modification by mPEG-succinimidyl carbonate (mPEG-SC) is a widely used method for N-terminal conjugation. In this study, we used it for site-directed modification on two ribosome-inactivating proteins (RIPs), alpha-momorcharin (α-MMC) and momordica anti-HIV protein (MAP30), from Momordica charantia L. According to the optimization of previous modification conditions, we compared Macro-Cap SP with SP-Sepharose FF chromatography for separating the final mPEGylated RIPs. Two kinds of methods both can obtain homogenous mPEGylated RIPs which were identified by sodium dodecylsulphate polyacrylamide gel electrophoresis (SDS-PAGE), isoelectric focusing electrophoresis (IEF), and matrix-assisted laser desorption ionization-time of flight/time of flight (MALDI-TOF/TOF) analysis. We also used iodine staining method to detect the amount of unmodified PEG. Furthermore, the inhibition activity of both mPEGylated and non-PEGylated RIPs against human lung adenocarcinoma epithelial A549 cells was detected. All of the results suggested that the mPEGylated α-MMC/MAP30 might be potentially developed as new anti-tumor drugs.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic , Lung Neoplasms/drug therapy , Momordica charantia/chemistry , Polyethylene Glycols/chemistry , Ribosome Inactivating Proteins, Type 2 , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/pharmacology
15.
Protein Expr Purif ; 111: 9-17, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25797209

ABSTRACT

MAP30 (Momordica Antiviral Protein 30 Kd), a single-stranded type-I ribosome inactivating protein, possesses versatile biological activities including anti-tumor abilities. However, the low efficiency penetrating into tumor cells hampers the tumoricidal effect of MAP30. This paper describes MAP30 fused with a human-derived cell penetrating peptide HBD which overcome the low uptake efficiency by tumor cells and exhibits higher anti-tumor bioactivity. MAP30 gene was cloned from the genomic DNA of Momordica charantia and the recombinant plasmid pET28b-MAP30-HBD was established and transferred into Escherichia coli BL21 (DE3). The recombinant MAP30-HBD protein (rMAP30-HBD) was expressed in a soluble form after being induced by 0.5mM IPTG for 14h at 15°C. The recombinant protein was purified to greater than 95% purity with Ni-NTA affinity chromatography. The rMAP30-HBD protein not only has topological inactivation and protein translation inhibition activity but also showed significant improvements in cytotoxic activity compared to that of the rMAP30 protein without HBD in the tested tumor cell lines, and induced higher apoptosis rates in HeLa cells analyzed by Annexin V-FITC with FACS. This paper demonstrated a new method for improving MAP30 protein anti-tumor activity and might have potential applications in cancer therapy area.


Subject(s)
Antineoplastic Agents , Cell-Penetrating Peptides , Neoplasms/drug therapy , Ribosome Inactivating Proteins, Type 2 , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/isolation & purification , Cell-Penetrating Peptides/pharmacology , HeLa Cells , Humans , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/biosynthesis , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/genetics , Ribosome Inactivating Proteins, Type 2/isolation & purification , Ribosome Inactivating Proteins, Type 2/pharmacology
16.
Biochem Biophys Res Commun ; 453(1): 117-23, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25261720

ABSTRACT

Aralin from Aralia elata is a newly identified type II ribosome- inactivating protein, which preferentially induces apoptosis in cancer cells. In this study, we identified that the aralin receptor is a 110-kDa high-density lipoprotein-binding protein (HDLBP), which functions as a HDL receptor. The sensitivities of tumor cell lines to aralin were dependent on the expression levels of the 110-kDa HDLBP and its forced expression in aralin-resistant Huh7 cells conferred aralin sensitivity. HDLBP-knockdown HeLa cells showed a significant aralin resistance in vitro and in vivo. Conversely, ectopic expression of the 150-kDa HDLBP resulted in increased aralin sensitivity in vivo, accompanying enhanced expression of the 110-kDa HDLBP. Thus, these results showed that the 110-kDa HDLBP in lipid rafts acted as an aralin receptor and that its expression levels determined aralin sensitivity, suggesting that aralin could be a promising anticancer drug for HDLBP-overexpressing tumors.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , RNA-Binding Proteins/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Aralia/chemistry , Cell Line, Tumor , Gene Knockdown Techniques , HeLa Cells , Hep G2 Cells , Humans , Lipoproteins, HDL/antagonists & inhibitors , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Membrane Microdomains/metabolism , Mice, Nude , Molecular Weight , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Receptors, Lipoprotein/antagonists & inhibitors , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/pharmacokinetics , Xenograft Model Antitumor Assays
17.
Toxicon ; 229: 107129, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37086901

ABSTRACT

Plant-derived triterpenoid saponins have been shown to play a powerful role in enhancing the cytotoxic activity of protein therapeutics. However, the mechanism of how saponins are acting is not clearly understood. In this study, momordin Ic (MIC), a triterpenoid saponin derived from Kochia scoparia (L.) Schrad., specifically enhance the antiproliferative effect of recombinant MAP30 (a type I ribosome inactivating protein, RIP) in breast cancer cells. Subsequently, the possible mechanism of how MIC enhanced the cytotoxicity of MAP30 was analyzed in detail. We observed the level of intracellular labeled MAP30 using fluorescence microscopy and flow cytometry. And a reporter protein, GAL9, was used to monitor the role of MIC in promoting endosomal escape. We found endosomal escape does not play a role for the enhancer effect of MIC while the effect of MIC on MAP30 is cholesterol dependent and that ganglioside GM1, a lipid raft marker, can competitively inhibit cytotoxicity of MAP30 enhanced by MIC. Finally, we provided some insights into the correlation between the sugar side chain of MIC and its role in enhancing of RIP cytotoxicity and altering of drug cell tropism.


Subject(s)
Antineoplastic Agents , Saponins , Triterpenes , G(M1) Ganglioside/pharmacology , Recombinant Proteins , Saponins/pharmacology , Cholesterol , Triterpenes/pharmacology , Ribosome Inactivating Proteins, Type 2/pharmacology
18.
Biomolecules ; 12(2)2022 02 11.
Article in English | MEDLINE | ID: mdl-35204796

ABSTRACT

Structurally similar catalytic subunits A of ricin (RTA) and viscumin (MLA) exhibit cytotoxic activity through ribosome inactivation. Ricin is more cytotoxic than viscumin, although the molecular mechanisms behind this difference are still poorly understood. To shed more light on this problem, we used a combined biochemical/molecular modeling approach to assess possible relationships between the activity of toxins and their structural/dynamic properties. Based on bioassay measurements, it was suggested that the differences in activity are associated with the ability of RTA and MLA to undergo structural/hydrophobic rearrangements during trafficking through the endoplasmic reticulum (ER) membrane. Molecular dynamics simulations and surface hydrophobicity mapping of both proteins in different media showed that RTA rearranges its structure in a membrane-like environment much more efficiently than MLA. Their refolded states also drastically differ in terms of hydrophobic organization. We assume that the higher conformational plasticity of RTA is favorable for the ER-mediated translocation pathway, which leads to a higher rate of toxin penetration into the cytoplasm.


Subject(s)
Ricin , Toxins, Biological , Endoplasmic Reticulum , Ribosome Inactivating Proteins, Type 2/pharmacology , Ricin/toxicity
19.
Acta Biochim Biophys Sin (Shanghai) ; 43(1): 52-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21148191

ABSTRACT

The effects of cytotoxic lectins, modeccin and phytohemagglutinin (PHA) on mouse macrophage cell line RAW264.7 was studied by detecting the induction of inflammatory mediators. Results showed that modeccin induced the release of tumor necrosis factor-α (TNF-α) from RAW264.7 cells with a bell-shape concentration-dependent profile. PHA that showed no significant cytotoxicity on RAW264.7 cells up to 100,000 ng/ml induced much higher level of TNF-α than modeccin. PHA simultaneously induced the secretion of granulocyte colony stimulation factor (G-CSF) from RAW264.7 cells with even much higher level than that of TNF-α, whereas modeccin did not. Furthermore, PHA induced the secretion of nitric oxide (NO) in RAW264.7 cells, while no significant level of NO was detected in the modeccin-treated cells. NH4Cl (a lysomotoropic agent) and cycloheximide (a ribosome inhibitor) strongly inhibited modeccin-induced TNF-α secretion, but no significant inhibitory effects of these reagents on the PHA-induced TNF-α secretion were observed. Contrary to modeccin-induced TNF-α secretion, even slightly increased TNF-α secretion was observed in PHA-treated cells in the presence of 10 mM NH4Cl. In addition, the inhibition profiles of modeccin-induced TNF-α secretion by various kinase inhibitors were different from those of PHA. These results suggested that the action mode of modeccin to stimulate RAW264.7 cells leading to the secretion of inflammatory molecules, including TNF-α, is distinct from that of PHA. On the other hand, significantly increased translocation of activator protein-1 (AP-1), a crucial transcription factor involved in expression of inflammatory molecules, into nucleus was observed in RAW264.7 cells treated with PHA and modeccin.


Subject(s)
Cytokines/metabolism , Nitric Oxide/metabolism , Phytohemagglutinins/pharmacology , Ribosome Inactivating Proteins, Type 2/pharmacology , Animals , Cell Line , Cell Nucleus/metabolism , Cycloheximide/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide Synthase Type II/biosynthesis , Protein Transport/drug effects , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Toxins (Basel) ; 13(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33499082

ABSTRACT

Ribosome-inactivating proteins (RIPs) are plant toxins that irreversibly damage ribosomes and other substrates, thus causing cell death. RIPs are classified in type 1 RIPs, single-chain enzymatic proteins, and type 2 RIPs, consisting of active A chains, similar to type 1 RIPs, linked to lectin B chains, which enable the rapid internalization of the toxin into the cell. For this reason, many type 2 RIPs are very cytotoxic, ricin, volkensin and stenodactylin being the most toxic ones. From the caudex of Adenia kirkii (Mast.) Engl., a new type 2 RIP, named kirkiin, was purified by affinity chromatography on acid-treated Sepharose CL-6B and gel filtration. The lectin, with molecular weight of about 58 kDa, agglutinated erythrocytes and inhibited protein synthesis in a cell-free system at very low concentrations. Moreover, kirkiin was able to depurinate mammalian and yeast ribosomes, but it showed little or no activity on other nucleotide substrates. In neuroblastoma cells, kirkiin inhibited protein synthesis and induced apoptosis at doses in the pM range. The biological characteristics of kirkiin make this protein a potential candidate for several experimental pharmacological applications both alone for local treatments and as component of immunoconjugates for systemic targeting in neurodegenerative studies and cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neuroblastoma/drug therapy , Passifloraceae/enzymology , Protein Synthesis Inhibitors/pharmacology , Ribosome Inactivating Proteins, Type 2/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Erythrocyte Aggregation/drug effects , Humans , Molecular Weight , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/isolation & purification , Protein Synthesis Inhibitors/toxicity , Ribosome Inactivating Proteins, Type 2/isolation & purification , Ribosome Inactivating Proteins, Type 2/toxicity , Ribosomes/drug effects , Ribosomes/genetics , Ribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL